1
|
Szopa IM, Majchrzak-Kuligowska K, Pingwara R, Kulka M, Taşdemir M, Gajewska M. A New Method of Canine CD4 + T Lymphocyte Differentiation Towards the Th17 Phenotype with Analysis of Properties and Mitochondrial Activity. Int J Mol Sci 2025; 26:4946. [PMID: 40430086 DOI: 10.3390/ijms26104946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Th17 lymphocytes are a distinct subpopulation of T cells that are characterized by the production of interleukins IL-17, IL-21, IL-22, and IL-26, and high expression of RORγt. These cells play an important role in inflammation and autoimmune diseases. Recent studies using rodent and human models have also highlighted their promising properties as agents in cellular immunotherapy for cancer. However, much less is known about the properties of canine Th17 lymphocytes, despite the domestic dog being an important model used in comparative medicine. In this study, we developed methods of activation and differentiation of canine CD4+ T lymphocytes towards the Th17 phenotype. Additionally, we targeted the Wnt/β-catenin signaling pathway to modulate the efficiency of Th17 cells differentiation. CD4+ T cells were successfully activated with magnetic EpoxyBeads, and in combination with the appropriate programming medium, they acquired the Th17 phenotype. Furthermore, indomethacin, an inhibitor of the Wnt/β-catenin pathway, significantly increased the efficiency of differentiation, causing elevated production of IL-17 and changed T cell metabolism by promoting oxidative phosphorylation. The protocol elaborated in our study provides an efficient method of canine Th17 lymphocyte differentiation. Our findings also suggested that the modification of the Wnt/β-catenin signaling pathway could be a valuable strategy for optimizing canine Th17 cell differentiation and advancing cell-based immunotherapy.
Collapse
Affiliation(s)
- Iwona Monika Szopa
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Kinga Majchrzak-Kuligowska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Rafał Pingwara
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Marek Kulka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Monika Taşdemir
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| |
Collapse
|
2
|
Ok SM, Jo JH, Cho HJ, Jang SM. RepID depletion enhances TWS119-induced erythropoiesis through chromatin reprogramming and transcription factor recruitment. Genes Genomics 2025; 47:533-540. [PMID: 40100582 DOI: 10.1007/s13258-025-01627-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Erythrocytes, derived from hematopoietic stem cells, are essential for oxygen transport, ensuring survival in all vertebrate animals. The process of erythropoiesis is associated with gene expression changes, but many key regulatory factors that govern erythroid differentiation remain to be fully understood. OBJECTIVE This study investigates the role of TWS119, a known GSK3β inhibitor, in inducing erythropoiesis in K562 erythroleukemia cells and explores the impact of Replication initiation determinant protein (RepID) depletion on the process. METHODS K562 cells were treated with TWS119 and erythropoiesis markers including various erythrocytic phenotypes were assessed. Chromatin-immunoprecipitation analysis was employed to examine the changes in chromatin structure and gene expression regulation. The impact of RepID depletion on TWS119-induced erythropoiesis was also evaluated by analyzing globin promoter euchromatinization and NRF2 binding. RESULTS TWS119 treatment led to erythrocytic phenotypes in K562 cells, such as red pellet formation, enucleation, and nucleus condensation, along with the upregulation of erythropoiesis markers. Furthermore, RepID depletion accelerated TWS119-mediated erythropoiesis. Chromatin-immunoprecipitation analysis revealed euchromatinization of the globin promoter and enhanced NRF2 binding in RepID-depleted cells, suggesting a mechanism of gene expression regulation during erythropoiesis. CONCLUSION This study demonstrates that TWS119 can induce erythropoiesis in K562 cells, and that RepID depletion enhances this process by modulating chromatin structure and facilitating transcription factor binding. These findings highlight a RepID-dependent mechanism in the regulation of gene expression during erythropoiesis.
Collapse
Affiliation(s)
- Seon-Mi Ok
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jae-Hyun Jo
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Hyo Je Cho
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Sang-Min Jang
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
3
|
Liang J, Yu M, Li Y, Zhao L, Wei Q. Glycogen synthase kinase-3: A potential immunotherapeutic target in tumor microenvironment. Biomed Pharmacother 2024; 173:116377. [PMID: 38442671 DOI: 10.1016/j.biopha.2024.116377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024] Open
Abstract
Glycogen synthase kinase-3(GSK-3) is a protein kinase that can phosphorylate over a hundred substrates and regulate cell differentiation, proliferation, and death. Researchers have acknowledged the pivotal role of abnormal activation of GSK-3 in the progression of various diseases over the past few decades. Recent studies have mostly concentrated on investigating the function of GSK-3 in the tumor microenvironment, specifically examining the interaction between TAM, NK cells, B cells, and T cells. Furthermore, GSK-3 exhibits a strong association with immunological checkpoints, such as programmed cell death protein 1. Novel GSK-3 inhibitors have potential in tumor immunotherapy, exerting beneficial effects on hematologic diseases and solid tumors. Nevertheless, there is a lack of reviews about the correlation between tumor-associated immune cells and GSK-3. This study intends to analyze the function and mechanism of GSK-3 comprehensively and systematically in the tumor microenvironment, with a special focus on its influence on various immune cells. The objective is to present novel perspectives for GSK-3 immunotherapy.
Collapse
Affiliation(s)
- Jingyi Liang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Meng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| |
Collapse
|
4
|
Wang Y, Wang R, Zhu J, Chen L. Identification of mitophagy and ferroptosis-related hub genes associated with intracerebral haemorrhage through bioinformatics analysis. Ann Hum Biol 2024; 51:2334719. [PMID: 38863372 DOI: 10.1080/03014460.2024.2334719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/21/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Mitophagy and ferroptosis occur in intracerebral haemorrhage (ICH) but our understanding of mitophagy and ferroptosis-related genes remains incomplete. AIM This study aims to identify shared ICH genes for both processes. METHODS ICH differentially expressed mitophagy and ferroptosis-related genes (DEMFRGs) were sourced from the GEO database and literature. Enrichment analysis elucidated functions. Hub genes were selected via STRING, MCODE, and MCC algorithms in Cytoscape. miRNAs targeting hubs were predicted using miRWalk 3.0, forming a miRNA-hub gene network. Immune microenvironment variances were assessed with MCP and TIMER. Potential small molecules for ICH were forecasted via CMap database. RESULTS 64 DEMFRGs and ten hub genes potentially involved in various processes like ferroptosis, TNF signalling pathway, MAPK signalling pathway, and NF-kappa B signalling pathway were discovered. Several miRNAs were identified as shared targets of hub genes. The ICH group showed increased infiltration of monocytic lineage and myeloid dendritic cells compared to the Healthy group. Ten potential small molecule drugs (e.g. Zebularine, TWS-119, CG-930) were predicted via CMap. CONCLUSION Several shared genes between mitophagy and ferroptosis potentially drive ICH progression via TNF, MAPK, and NF-kappa B pathways. These results offer valuable insights for further exploring the connection between mitophagy, ferroptosis, and ICH.
Collapse
Affiliation(s)
- Yan Wang
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Rufeng Wang
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Jianzhong Zhu
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Ling Chen
- Department of Gynaecology, People's Hospital Affiliated to Cangzhou Medical College, Cangzhou, China
| |
Collapse
|
5
|
Kori M, Temiz K, Gov E. Network medicine approaches for identification of novel prognostic systems biomarkers and drug candidates for papillary thyroid carcinoma. J Cell Mol Med 2023; 27:4171-4180. [PMID: 37859510 PMCID: PMC10746936 DOI: 10.1111/jcmm.18002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is one of the most common endocrine carcinomas worldwide and the aetiology of this cancer is still not well understood. Therefore, it remains important to understand the disease mechanism and find prognostic biomarkers and/or drug candidates for PTC. Compared with approaches based on single-gene assessment, network medicine analysis offers great promise to address this need. Accordingly, in the present study, we performed differential co-expressed network analysis using five transcriptome datasets in patients with PTC and healthy controls. Following meta-analysis of the transcriptome datasets, we uncovered common differentially expressed genes (DEGs) for PTC and, using these genes as proxies, found a highly clustered differentially expressed co-expressed module: a 'PTC-module'. Using independent data, we demonstrated the high prognostic capacity of the PTC-module and designated this module as a prognostic systems biomarker. In addition, using the nodes of the PTC-module, we performed drug repurposing and text mining analyzes to identify novel drug candidates for the disease. We performed molecular docking simulations, and identified: 4-demethoxydaunorubicin hydrochloride, AS605240, BRD-A60245366, ER 27319 maleate, sinensetin, and TWS119 as novel drug candidates whose efficacy was also confirmed by in silico analyzes. Consequently, we have highlighted here the need for differential co-expression analysis to gain a systems-level understanding of a complex disease, and we provide candidate prognostic systems biomarker and novel drugs for PTC.
Collapse
Affiliation(s)
- Medi Kori
- Faculty of Health SciencesAcibadem Mehmet Ali Aydinlar UniversityİstanbulTürkiye
- Department of BioengineeringMarmara UniversityİstanbulTürkiye
| | - Kubra Temiz
- Department of BioengineeringAdana Alparslan Turkes Science and Technology UniversityAdanaTürkiye
| | - Esra Gov
- Department of BioengineeringAdana Alparslan Turkes Science and Technology UniversityAdanaTürkiye
| |
Collapse
|
6
|
Feng YB, Chen L, Chen FX, Yang Y, Chen GH, Zhou ZH, Xu CF. Immunopotentiation effects of apigenin on NK cell proliferation and killing pancreatic cancer cells. Int J Immunopathol Pharmacol 2023; 37:3946320231161174. [PMID: 36848930 PMCID: PMC9974612 DOI: 10.1177/03946320231161174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Apigenin is a kind of flavonoid with many beneficial biological effects. It not only has direct cytotoxicity to tumor cells, but also can boost the antitumor effect of immune cells by modulating immune system. The purpose of this study was to investigate the proliferation of NK cells treated with apigenin and its cytotoxicity to pancreatic cancer cells in vitro, and explore its potential molecular mechanism. In this study, the effect of apigenin on NK cell proliferation and killing pancreatic cancer cells were measured by CCK-8 assay. Perforin, granzyme B (Gran B), CD107a, and NKG2D expressions of NK cells induced with apigenin were detected by flow cytometry (FCM). The mRNA expression of Bcl-2, Bax and protein expression of Bcl-2, Bax, p-ERK, and p-JNK in NK cells were evaluated by qRT-PCR and western blotting analysis, respectively. The results showed that appropriate concentration of apigenin could significantly promote the proliferation of NK cells in vitro and enhance the killing activity of NK cells against pancreatic cancer cells. The expressions of surface antigen NKG2D and intracellular antigen perforin and Gran B of NK cells were upregulated after treating with apigenin. Bcl-2 mRNA expression was increased, while Bax mRNA expression was decreased. Similarly, the expression of Bcl-2, p-JNK, and p-ERK protein was upregulated, and the expression of Bax protein was downregulated. The molecular mechanism of the immunopotentiation effects of apigenin may be that it up-regulates Bcl-2 and down-regulates Bax expression at the gene and protein levels to facilitate NK cell proliferation, and up-regulates the expression of perforin, Gran B, and NKG2D through the activation of JNK and ERK pathways to enhance NK cell cytotoxicity.
Collapse
Affiliation(s)
- Yong-Bo Feng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China,Department of Gastroenterology, Nanjing Gaochun Hospital of Chinese Medicine, Jiangsu, China
| | - Ling Chen
- Department of Central Laboratory, the 71st Group Army Hospital of PLA Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, Jiangsu, China
| | - Fu-Xing Chen
- Department of Central Laboratory, the 71st Group Army Hospital of PLA Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, Jiangsu, China
| | - Yang Yang
- Department of Pharmacy, the 71st Group Army Hospital of PLA Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, Jiangsu, China
| | - Guo-Hua Chen
- Department of General Surgery, Nanjing Gaochun Hospital of Chinese Medicine, Jiangsu, China
| | - Zhong-Hai Zhou
- Department of Central Laboratory, the 71st Group Army Hospital of PLA Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, Jiangsu, China,Zhong-Hai Zhou, Department of Central Laboratory, the 71st Group Army Hospital of PLA Army, The Affiliated Huaihai Hospital of Xuzhou Medical University, 236 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Chun-Fang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China,Chun-Fang Xu, Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi street, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
7
|
Corsale AM, Di Simone M, Lo Presti E, Dieli F, Meraviglia S. γδ T cells and their clinical application in colon cancer. Front Immunol 2023; 14:1098847. [PMID: 36793708 PMCID: PMC9923022 DOI: 10.3389/fimmu.2023.1098847] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
In recent years, research has focused on colorectal cancer to implement modern treatment approaches to improve patient survival. In this new era, γδ T cells constitute a new and promising candidate to treat many types of cancer because of their potent killing activity and their ability to recognize tumor antigens independently of HLA molecules. Here, we focus on the roles that γδ T cells play in antitumor immunity, especially in colorectal cancer. Furthermore, we provide an overview of small-scale clinical trials in patients with colorectal cancer employing either in vivo activation or adoptive transfer of ex vivo expanded γδ T cells and suggest possible combinatorial approaches to treat colon cancer.
Collapse
Affiliation(s)
- Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR)I, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| |
Collapse
|
8
|
Bustos X, Snedal S, Tordesillas L, Pelle E, Abate-Daga D. γδ T Cell-Based Adoptive Cell Therapies Against Solid Epithelial Tumors. Cancer J 2022; 28:270-277. [PMID: 35880936 PMCID: PMC9335899 DOI: 10.1097/ppo.0000000000000606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ABSTRACT Conventionally, adoptive cell therapies have been developed and optimized using αβ T cells. However, the understudied and less abundant γδ T cells offer unique advantages to the immunotherapy field especially for therapies against solid tumors. Recently, γδ T-cell potential against a broad spectrum of malignant cells has been demonstrated in the preclinical setting. In the clinic, γδ T-cell-based immunotherapies have proven to be safe; however, their efficacy needs improvement. Considering the growing body of literature reflecting the increasing interest in γδ T cells, we sought to capture the current topics of discussion in the field, pertaining to their use in adoptive immunotherapy. We aimed to compile information about γδ T-cell enhancement in terms of expansion, phenotype, and inhibitory receptors, in addition to the latest advances in preclinical and clinical research using γδ T cells specifically against solid epithelial tumors.
Collapse
|
9
|
Nel AE, Mei KC, Liao YP, Lu X. Multifunctional Lipid Bilayer Nanocarriers for Cancer Immunotherapy in Heterogeneous Tumor Microenvironments, Combining Immunogenic Cell Death Stimuli with Immune Modulatory Drugs. ACS NANO 2022; 16:5184-5232. [PMID: 35348320 PMCID: PMC9519818 DOI: 10.1021/acsnano.2c01252] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In addition to the contribution of cancer cells, the solid tumor microenvironment (TME) has a critical role in determining tumor expansion, antitumor immunity, and the response to immunotherapy. Understanding the details of the complex interplay between cancer cells and components of the TME provides an unprecedented opportunity to explore combination therapy for intervening in the immune landscape to improve immunotherapy outcome. One approach is the introduction of multifunctional nanocarriers, capable of delivering drug combinations that provide immunogenic stimuli for improvement of tumor antigen presentation, contemporaneous with the delivery of coformulated drug or synthetic molecules that provide immune danger signals or interfere in immune-escape, immune-suppressive, and T-cell exclusion pathways. This forward-looking review will discuss the use of lipid-bilayer-encapsulated liposomes and mesoporous silica nanoparticles for combination immunotherapy of the heterogeneous immune landscapes in pancreatic ductal adenocarcinoma and triple-negative breast cancer. We describe how the combination of remote drug loading and lipid bilayer encapsulation is used for the synthesis of synergistic drug combinations that induce immunogenic cell death, interfere in the PD-1/PD-L1 axis, inhibit the indoleamine-pyrrole 2,3-dioxygenase (IDO-1) immune metabolic pathway, restore spatial access to activated T-cells to the cancer site, or reduce the impact of immunosuppressive stromal components. We show how an integration of current knowledge and future discovery can be used for a rational approach to nanoenabled cancer immunotherapy.
Collapse
Affiliation(s)
- André E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California 90095, United States
| | - Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Lu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
10
|
Xu J, Li Y, Fan Q, Shu Y, Yang L, Cui T, Gu K, Tao M, Wang X, Cui C, Xu N, Xiao J, Gao Q, Liu Y, Zhang T, Bai Y, Li W, Zhang Y, Dai G, Ma D, Zhang J, Bai C, Huang Y, Liao W, Wu L, Chen X, Yang Y, Wang J, Ji S, Zhou H, Wang Y, Ma Z, Wang Y, Peng B, Sun J, Mancao C. Clinical and biomarker analyses of sintilimab versus chemotherapy as second-line therapy for advanced or metastatic esophageal squamous cell carcinoma: a randomized, open-label phase 2 study (ORIENT-2). Nat Commun 2022; 13:857. [PMID: 35165274 PMCID: PMC8844279 DOI: 10.1038/s41467-022-28408-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
This randomized, open-label, multi-center phase 2 study (NCT03116152) assessed sintilimab, a PD-1 inhibitor, versus chemotherapy in patients with esophageal squamous cell carcinoma after first-line chemotherapy. The primary endpoint was overall survival (OS), while exploratory endpoint was the association of biomarkers with efficacy. The median OS in the sintilimab group was significantly improved compared with the chemotherapy group (median OS 7.2 vs.6.2 months; P = 0.032; HR = 0.70; 95% CI, 0.50-0.97). Incidence of treatment-related adverse events of grade 3-5 was lower with sintilimab than with chemotherapy (20.2 vs. 39.1%). Patients with high T-cell receptor (TCR) clonality and low molecular tumor burden index (mTBI) showed the longest median OS (15.0 months). Patients with NLR < 3 at 6 weeks post-treatment had a significantly prolonged median OS (16.6 months) compared with NLR ≥ 3. The results demonstrate a significant improvement in OS of sintilimab compared to chemotherapy as second-line treatment for advanced or metastatic ESCC.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Yi Li
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Yang
- Department of Radiotherapy, Nantong Tumor Hospital, Nantong, China
| | - Tongjian Cui
- Department of Medical Oncology, Fujian Provincial Hospital, Fuzhou, China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiuwen Wang
- Department of Chemotherapy, Qilu Hospital of Shandong University, Jinan, China
| | - Chengxu Cui
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Juxiang Xiao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Quanli Gao
- Department of Immunotherapy, Henan Cancer Hospital, Zhengzhou, China
| | - Yunpeng Liu
- Department of Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Tao Zhang
- Cancer Center, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yiping Zhang
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Guanghai Dai
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Dong Ma
- Department of Gastrointestinal Oncology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Jingdong Zhang
- Department of Gastroenterology, Liaoning Cancer Hospital, Shenyang, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Yunnan Cancer Hospital, Kunming, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Wu
- Departmentof Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha, China
| | - Xi Chen
- Department of Oncology, No. 900 Hospital of The Joint Logistic Support Force, Fuzhou, China
| | - Yan Yang
- Department of Gastrointestinal Oncology, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Junye Wang
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Shoujian Ji
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui Zhou
- Innovent Biologics, Inc., Suzhou, China
| | - Yan Wang
- Innovent Biologics, Inc., Suzhou, China
| | - Zhuo Ma
- Innovent Biologics, Inc., Suzhou, China
| | | | - Bo Peng
- Innovent Biologics, Inc., Suzhou, China
| | - Jiya Sun
- Innovent Biologics, Inc., Suzhou, China
| | | |
Collapse
|
11
|
Raineri F, Bourgoin-Voillard S, Cossutta M, Habert D, Ponzo M, Houppe C, Vallée B, Boniotto M, Chalabi-Dchar M, Bouvet P, Couvelard A, Cros J, Debesset A, Cohen JL, Courty J, Cascone I. Nucleolin Targeting by N6L Inhibits Wnt/β-Catenin Pathway Activation in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13122986. [PMID: 34203710 PMCID: PMC8232280 DOI: 10.3390/cancers13122986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and resistant cancer with no available effective therapy. We have previously demonstrated that nucleolin targeting by N6L impairs tumor growth and normalizes tumor vessels in PDAC mouse models. Here, we investigated new pathways that are regulated by nucleolin in PDAC. We found that N6L and nucleolin interact with β-catenin. We found that the Wnt/β-catenin pathway is activated in PDAC and is necessary for tumor-derived 3D growth. N6L and nucleolin loss of function induced by siRNA inhibited Wnt pathway activation by preventing β-catenin stabilization in PDAC cells. N6L also inhibited the growth and the activation of the Wnt/β-catenin pathway in vivo in mice and in 3D cultures derived from MIA PaCa2 tumors. On the other hand, nucleolin overexpression increased β-catenin stabilization. In conclusion, in this study, we identified β-catenin as a new nucleolin interactor and suggest that the Wnt/β-catenin pathway could be a new target of the nucleolin antagonist N6L in PDAC.
Collapse
Affiliation(s)
- Fabio Raineri
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Sandrine Bourgoin-Voillard
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
- University of Grenoble Alpes, CNRS, Grenoble INP, Inserm U1055, LBFA and BEeSy, PROMETHEE Proteomic Platform, 38400 Saint-Martin d’Heres, France
- University of Grenoble Alpes, CNRS, Grenoble INP, TIMC, PROMETHEE Proteomic Platform, 38000 Grenoble, France
- CHU Grenoble Alpes, Institut de Biologie et de Pathologie, 38043 Grenoble, France
| | - Mélissande Cossutta
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapie, 94010 Créteil, France
| | - Damien Habert
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Matteo Ponzo
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Claire Houppe
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Benoît Vallée
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Michele Boniotto
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - Mounira Chalabi-Dchar
- Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, University of Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 69008 Lyon, France; (M.C.-D.); (P.B.)
| | - Philippe Bouvet
- Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, University of Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 69008 Lyon, France; (M.C.-D.); (P.B.)
- University of Lyon, Ecole Normale Supérieure de Lyon, 69342 Lyon, France
| | - Anne Couvelard
- Département de Pathologie, Hôpital Bichat APHP DHU UNITY, 75018 Paris, France; (A.C.); (J.C.)
| | - Jerome Cros
- Département de Pathologie, Hôpital Bichat APHP DHU UNITY, 75018 Paris, France; (A.C.); (J.C.)
| | - Anais Debesset
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
| | - José L. Cohen
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapie, 94010 Créteil, France
| | - José Courty
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapie, 94010 Créteil, France
| | - Ilaria Cascone
- University Paris Est Créteil, INSERM, IMRB, 94010 Créteil, France; (F.R.); (S.B.-V.); (M.C.); (D.H.); (M.P.); (C.H.); (B.V.); (M.B.); (A.D.); (J.L.C.); (J.C.)
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapie, 94010 Créteil, France
- Correspondence: ; Tel.: +33-149-813-765
| |
Collapse
|
12
|
Sabbaghi A, Miri SM, Keshavarz M, Mahooti M, Zebardast A, Ghaemi A. Role of γδ T cells in controlling viral infections with a focus on influenza virus: implications for designing novel therapeutic approaches. Virol J 2020; 17:174. [PMID: 33183352 PMCID: PMC7659406 DOI: 10.1186/s12985-020-01449-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Influenza virus infection is among the most detrimental threats to the health of humans and some animals, infecting millions of people annually all around the world and in many thousands of cases giving rise to pneumonia and death. All those health crises happen despite previous and recent developments in anti-influenza vaccination, suggesting the need for employing more sophisticated methods to control this malign infection. Main body The innate immunity modules are at the forefront of combating against influenza infection in the respiratory tract, among which, innate T cells, particularly gamma-delta (γδ) T cells, play a critical role in filling the gap needed for adaptive immune cells maturation, linking the innate and adaptive immunity together. Upon infection with influenza virus, production of cytokines and chemokines including CCL3, CCL4, and CCL5 from respiratory epithelium recruits γδ T cells at the site of infection in a CCR5 receptor-dependent fashion. Next, γδ T cells become activated in response to influenza virus infection and produce large amounts of proinflammatory cytokines, especially IL-17A. Regardless of γδ T cells' roles in triggering the adaptive arm of the immune system, they also protect the respiratory epithelium by cytolytic and non-cytolytic antiviral mechanisms, as well as by enhancing neutrophils and natural killer cells recruitment to the infection site. CONCLUSION In this review, we explored varied strategies of γδ T cells in defense to influenza virus infection and how they can potentially provide balanced protective immune responses against infected cells. The results may provide a potential window for the incorporation of intact or engineered γδ T cells for developing novel antiviral approaches or for immunotherapeutic purposes.
Collapse
Affiliation(s)
- Ailar Sabbaghi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Seyed Mohammad Miri
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mehran Mahooti
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Arghavan Zebardast
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran.
| |
Collapse
|
13
|
Xu D, Li F, Hou K, Gou X, Fang W, Li Y. XQ-1H attenuates ischemic injury in PC12 cells via Wnt/β-catenin signaling though inhibition of apoptosis and promotion of proliferation. Cell Biol Int 2020; 44:2363-2369. [PMID: 32761926 DOI: 10.1002/cbin.11438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 08/01/2020] [Indexed: 01/10/2023]
Abstract
10-O-(N,N-dimethylaminoethyl)-ginkgolide B methanesulfonate (XQ-1H) is a new derivative of ginkgolide B and has previously been proven to exert neuroprotective effects on ischemic injury. However, it is not clear whether XQ-1H affects the cell survival and proliferation in oxygen-glucose deprivation/reoxygenation (OGD/R) damaged PC12 cells. Our results showed that OGD/R improved cell viability after 24 hr of posttreatment with XQ-1H (10 or 5 μM), inhibiting cell injury and apoptosis by upregulating the expression of brain-derived neurotrophic factor, nerve growth factor, and antiapoptotic B-cell lymphoma-extra large, while reducing proapoptotic cleaved caspase-3 protein. By introducing the Wnt/β-catenin signaling inhibitor XAV-939 and 5-bromo-2'-deoxyuridine staining, it was proved that XQ-1H promoted the proliferation of PC12 cells in a Wnt-signal-dependent manner via inhibiting the activation of glycogen synthase kinase-3β after phosphatidylinositol 3-kinase/protein kinase B signal activation, thereby activating Wnt1, β-catenin, and the expression of downstream neurogenic differentiation 1 and cyclin D1, which was comparable to Wnt/β-catenin signaling agonist 4,6-disubstituted pyrrolopyrimidine. We conclude that XQ-1H, after OGD/R damage to PC12 cells, may limit cell apoptosis in a Wnt/β-catenin signal-dependent manner, promoting cell proliferation and survival.
Collapse
Affiliation(s)
- Dan Xu
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fengyang Li
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Kai Hou
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xue Gou
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Weirong Fang
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yunman Li
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Xu D, Li F, Xue G, Hou K, Fang W, Li Y. Effect of Wnt signaling pathway on neurogenesis after cerebral ischemia and its therapeutic potential. Brain Res Bull 2020; 164:1-13. [PMID: 32763283 DOI: 10.1016/j.brainresbull.2020.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/08/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Neurogenesis process in the chronic phase of ischemic stroke has become the focus of research on stroke treatment recently, mainly through the activation of related pathways to increase the differentiation of neural stem cells (NSCs) in the brain sub-ventricular zone (SVZ) and subgranular zone (SGZ) of hippocampal dentate gyrus (DG) areas into neurons, promoting neurogenesis. While there is still debate about the longevity of active adult neurogenesis in humans, the SVZ and SGZ have the capacity to upregulate neurogenesis in response to cerebral ischemia, which opens discussion about potential treatment strategies to harness this neuronal regenerative response. Wnt signaling pathway is one of the most important approaches potentially targeting on neurogenesis after cerebral ischemia, appropriate activation of which in NSCs may help to improve the sequelae of cerebral ischemia. Various therapeutic approaches are explored on preclinical stage to target endogenous neurogenesis induced by Wnt signaling after stroke onset. This article describes the composition of Wnt signaling pathway and the process of neurogenesis after cerebral ischemia, and emphatically introduces the recent studies on the mechanisms of this pathway for post-stroke neurogenesis and the therapeutic possibility of activating the pathway to improve neurogenesis after stroke.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Gou Xue
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
15
|
Zhang Z, Lv Z, Zhang W, Guo M, Li C. A novel β-catenin from Apostichopus japonicus mediates Vibrio splendidus-induced inflammatory-like response. Int J Biol Macromol 2020; 156:730-739. [PMID: 32311399 DOI: 10.1016/j.ijbiomac.2020.04.103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 12/26/2022]
Abstract
β-catenin, an adaptor molecule in Wnt/β-catenin signaling pathway, is associated with different physiological processes such as intestinal immune, apoptosis, and inflammation-associated response. However, the function of β-catenin is still largely unknown in Apostichopus japonicus. In the present study, we cloned and characterized β-catenin gene from A. japonicus by RNA-seq and RACE approaches. The complete sequence of Ajβ-catenin consisted of a 5' UTR of 166 bp, a 3' UTR of 501 bp and an ORF of 2433 bp encoding a protein of 810 amino acids. Ajβ-catenin has a GSK-β consensus phosphorylation site of 21 amino acids located at N-terminal region and twelve Armadillo/β-catenin-like repeat (ARM) domains from 145 to 671 aa. Spatial expression analysis revealed that Ajβ-catenin mRNA levels displayed higher abundance in intestine. For Vibrio splendidus challenged sea cucumber, Ajβ-catenin transcripts reached their peak at 6 h and remained at higher levels until 24 h post infection in comparison with that of the control group. GSK-3β inhibitor treatment could induce both Ajβ-catenin and the inflammatory factors expression. Ajβ-catenin silencing could also down-regulate inflammatory factors expression. These results collectively suggested that Ajβ-catenin was a novel molecule mediate V. splendidus-induced immune response of A. japonicus via regulating the inflammatory factors expression.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Zhimeng Lv
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Weiwei Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China.
| |
Collapse
|
16
|
Lu H, Shi T, Wang M, Li X, Gu Y, Zhang X, Zhang G, Chen W. B7-H3 inhibits the IFN-γ-dependent cytotoxicity of Vγ9Vδ2 T cells against colon cancer cells. Oncoimmunology 2020; 9:1748991. [PMID: 32363121 PMCID: PMC7185217 DOI: 10.1080/2162402x.2020.1748991] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/12/2020] [Accepted: 03/22/2020] [Indexed: 12/19/2022] Open
Abstract
The immunoregulatory protein B7-H3, a member of the B7 family, has been confirmed to be highly expressed in colon cancer. However, the exact influence of B7-H3 on the features and antitumor ability of γδT cells in colon cancer remains unknown. In the present study, we investigated that the proportions of B7-H3+ γδT cells were distinctly increased in the peripheral blood and tumor tissues of colon cancer patients. B7-H3 blockade or knockdown promoted proliferation, inhibited cell apoptosis and induced the expression of activation markers (CD25 and CD69) on Vδ2 T cells. In contrast, treatment with the B7-H3 agonist 4H7 had the opposite effect. Furthermore, B7-H3 suppressed IFN-γ expression by inhibiting T-bet in Vδ2 T cells. Moreover, B7-H3 mediated the inhibition of Vδ2 T cell cytotoxicity via the downregulation of IFN-γ and perforin/granzyme B expression. More importantly, blocking the B7-H3 function significantly enhanced the cytotoxicity of Vδ2 T cells against colon cancer cells in vivo. Therefore, the inhibition or blockade of B7-H3 is a potential immunotherapeutic approach for colon cancer.
Collapse
Affiliation(s)
- Huimin Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Xiaomi Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Song D, Zhang X, Chen J, Liu X, Xue J, Zhang L, Lan X. Wnt canonical pathway activator TWS119 drives microglial anti-inflammatory activation and facilitates neurological recovery following experimental stroke. J Neuroinflammation 2019; 16:256. [PMID: 31810470 PMCID: PMC6896312 DOI: 10.1186/s12974-019-1660-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/26/2019] [Indexed: 11/29/2022] Open
Abstract
Background Ischemic stroke is a leading cause of disability worldwide and characteristically accompanied by downregulation of the Wnt/β-catenin signaling. Activation of Wnt/β-catenin signaling emerges to attenuate neuroinflammation after ischemic stroke; however, its effect on modulating microglial polarization is largely unknown. Here, we explored whether Wnt/β-catenin pathway activator TWS119 facilitated long-term neurological recovery via modulating microglia polarization after experimental stroke. Methods Ischemic stroke mice model was induced by permanent distal middle cerebral artery occlusion plus 1 h hypoxia. TWS119 was administrated from day 1 to 14 after stroke. Neurological deficits were monitored up to 21 days after stroke. Angiogenesis, neural plasticity, microglial polarization, and microglia-associated inflammatory cytokines were detected in the peri-infarct cortex at days 14 and 21 after stroke. Primary microglia and mouse brain microvascular endothelial cell lines were employed to explore the underlying mechanism in vitro. Results TWS119 mitigated neurological deficits at days 14 and 21 after experimental stroke, paralleled by acceleration on angiogenesis and neural plasticity in the peri-infarct cortex. Mechanistically, cerebral ischemia induced production of microglia-associated proinflammatory cytokines and priming of activated microglia toward pro-inflammatory polarization, whereas TWS119 ameliorated microglia-mediated neuroinflammatory status following ischemic stroke and promoted angiogenesis by modulating microglia to anti-inflammatory phenotype. The beneficial efficacy of TWS119 in microglial polarization was largely reversed by selective Wnt/β-catenin pathway blockade in vitro, suggesting that TWS119-enabled pro-inflammatory to anti-inflammatory phenotype switch of microglia was possibly mediated by Wnt/β-catenin signaling. Conclusions Wnt/β-catenin pathway activator TWS119 ameliorated neuroinflammatory microenvironment following chronic cerebral ischemia via modulating microglia towards anti-inflammatory phenotype, and facilitates neurological recovery in an anti-inflammatory phenotype polarization-dependent manner. Activation of Wnt/β-catenin pathway following ischemic stroke might be a potential restorative strategy targeting microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Degang Song
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Department of Neurology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China. .,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
| | - Junmin Chen
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaoxia Liu
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Jing Xue
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xifa Lan
- Department of Neurology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| |
Collapse
|
18
|
Li X, Lu H, Gu Y, Zhang X, Zhang G, Shi T, Chen W. Tim-3 suppresses the killing effect of Vγ9Vδ2 T cells on colon cancer cells by reducing perforin and granzyme B expression. Exp Cell Res 2019; 386:111719. [PMID: 31726050 DOI: 10.1016/j.yexcr.2019.111719] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/24/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023]
Abstract
Gamma delta (γδ) T cell-based tumor immunotherapy has been one of the most promising cancer immunotherapeutic strategies. However, the key regulators of the Vγ9Vδ2 T cell-mediated antitumor response remain unclear. Recently, mounting reports have indicated that Tim-3 performs critical roles in the regulation of the activities of immune cells, including Vγ9Vδ2 T cells. However, the roles of Tim-3 in Vγ9Vδ2 T cell-mediated killing of colon cancer cells and the underlying mechanism remain largely unknown. Here, the proportion of Tim-3+ γδ T cells was significantly increased in both the peripheral blood and colon cancer tissue of patients and was significantly associated with TNM staging and tumor volume. Additionally, the activation of Tim-3 signaling significantly inhibited the killing efficiency of Vγ9Vδ2 T cells against colon cancer cells. In addition, Tim-3 signaling reduced the expression of perforin and granzyme B in Vγ9Vδ2 T cells. Blocking the perforin/granzyme B pathway also decreased the cytotoxicity of Vγ9Vδ2 T cells to colon cancer cells. Moreover, Tim-3 signaling reduced the perforin and granzyme B expression of Vγ9Vδ2 T cells in an ERK1/2 signaling pathway-dependent manner. This knowledge reveals that Tim-3 may be a promising therapeutic target to improve Vγ9Vδ2 T cell-based adoptive immunotherapy for colon cancer.
Collapse
Affiliation(s)
- Xiaomi Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Huimin Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China; Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
| |
Collapse
|
19
|
Xu D, Hou K, Li F, Chen S, Fang W, Li Y. XQ-1H alleviates cerebral ischemia in mice through inhibition of apoptosis and promotion of neurogenesis in a Wnt/β-catenin signaling dependent way. Life Sci 2019; 235:116844. [PMID: 31499069 DOI: 10.1016/j.lfs.2019.116844] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 11/27/2022]
Abstract
AIMS 10-O-(N,N-dimethylaminoethyl)-ginkgolide B methanesulfonate (XQ-1H), a new derivative of ginkgolide B, has drawn great attention for its potent bioactivities against ischemia-induced injury. The purpose of this study was to further investigate the effect of XQ-1H against acute ischemic stroke by inducing middle cerebral artery occlusion/reperfusion (MCAO/R) injuries in mice. MAIN METHODS Treatment of XQ-1H (78 or 39 mg/kg, i.g., bid) 2 h after MCAO improved motor skills and ameliorated the severity of brain infarction and apoptosis seen in the mice by diminishing pathological changes and the activation of a pro-apoptotic protein Cleaved-Caspase-3, which in turn induced anti-apoptotic Bcl-xL. Through introducing Wnt/β-catenin signaling inhibitor XAV-939, XQ-1H was proven to intensively promoted neurogenesis in the peri-infarct cortex, subventricular area (SVZ) and the dentate gyrus (DG) subgranular area (SGZ) in a Wnt signal dependent way by compromising the activation of GSK3β, which in turn upregulated Wnt1, β-catenin, Neuro D1 and Cyclin D1, most possibly through the activation of PI3K/Akt signaling via the upregulation of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). KEY FINDINGS We conclude that XQ-1H preserved the motor functions, limited apoptosis, and concomitantly promoted neurogenesis-related protein expression by Wnt signaling-dependently compromising GSK3β/Caspase-3 activity and enhancing the expression of Wnt1/β-catenin/Neuro D1/Cyclin D1 and Bcl-xL. SIGNIFICANCE This research may benefit the development of stroke therapeutics targeting neurogenesis through Wnt upregulation by XQ-1H.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shijie Chen
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
20
|
Chen YQ, Li PC, Pan N, Gao R, Wen ZF, Zhang TY, Huang F, Wu FY, Ou XL, Zhang JP, Zhu XJ, Hu HM, Chen K, Cai YL, Wang LX. Tumor-released autophagosomes induces CD4 + T cell-mediated immunosuppression via a TLR2-IL-6 cascade. J Immunother Cancer 2019; 7:178. [PMID: 31300052 PMCID: PMC6625067 DOI: 10.1186/s40425-019-0646-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CD4+ T cells are critical effectors of anti-tumor immunity, but how tumor cells influence CD4+ T cell effector function is not fully understood. Tumor cell-released autophagosomes (TRAPs) are being recognized as critical modulators of host anti-tumor immunity during tumor progression. Here, we explored the mechanistic aspects of TRAPs in the modulation of CD4+ T cells in the tumor microenvironment. METHODS TRAPs isolated from tumor cell lines and pleural effusions or ascites of cancer patients were incubated with CD4+ T cells to examine the function and mechanism of TRAPs in CD4+ T cell differentiation and function. TRAPs-elicited CD4+ T cells were tested for their suppression of effector T cell function, induction of regulatory B cells, and promotion of tumorigenesis and metastasis in a mouse model. RESULTS Heat shock protein 90α (HSP90α) on the surface of TRAPs from malignant effusions of cancer patients and tumor cell lines stimulated CD4+ T cell production of IL-6 via a TLR2-MyD88-NF-κB signal cascade. TRAPs-induced autocrine IL-6 further promoted CD4+ T cells secretion of IL-10 and IL-21 via STAT3. Notably, TRAPs-elicited CD4+ T cells inhibited CD4+ and CD8+ effector T cell function in an IL-6- and IL-10-dependent manner and induced IL-10-producing regulatory B cells (Bregs) via IL-6, IL-10 and IL-21, thereby promoting tumor growth and metastasis. Consistently, inhibition of tumor autophagosome formation or IL-6 secretion by CD4+ T cells markedly retarded tumor growth. Furthermore, B cell or CD4+ T cell depletion impeded tumor growth by increasing effector T cell function. CONCLUSIONS HSP90α on the surface of TRAPs programs the immunosuppressive functions of CD4+ T cells to promote tumor growth and metastasis. TRAPs or their membrane-bound HSP90α represent important therapeutic targets to reverse cancer-associated immunosuppression and improve immunotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Chen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Peng-Cheng Li
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Rong Gao
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Fa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Tian-Yu Zhang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang Huang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang-Yuan Wu
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Xi-Long Ou
- Department of Gastroenterology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jin-Ping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xue-Jun Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Detroit, MI, 48201, USA.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yun-Lang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
21
|
Chen YQ, Li PC, Pan N, Gao R, Wen ZF, Zhang TY, Huang F, Wu FY, Ou XL, Zhang JP, Zhu XJ, Hu HM, Chen K, Cai YL, Wang LX. Tumor-released autophagosomes induces CD4 + T cell-mediated immunosuppression via a TLR2-IL-6 cascade. J Immunother Cancer 2019. [PMID: 31300052 DOI: 10.1186/s40425-019-0646-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CD4+ T cells are critical effectors of anti-tumor immunity, but how tumor cells influence CD4+ T cell effector function is not fully understood. Tumor cell-released autophagosomes (TRAPs) are being recognized as critical modulators of host anti-tumor immunity during tumor progression. Here, we explored the mechanistic aspects of TRAPs in the modulation of CD4+ T cells in the tumor microenvironment. METHODS TRAPs isolated from tumor cell lines and pleural effusions or ascites of cancer patients were incubated with CD4+ T cells to examine the function and mechanism of TRAPs in CD4+ T cell differentiation and function. TRAPs-elicited CD4+ T cells were tested for their suppression of effector T cell function, induction of regulatory B cells, and promotion of tumorigenesis and metastasis in a mouse model. RESULTS Heat shock protein 90α (HSP90α) on the surface of TRAPs from malignant effusions of cancer patients and tumor cell lines stimulated CD4+ T cell production of IL-6 via a TLR2-MyD88-NF-κB signal cascade. TRAPs-induced autocrine IL-6 further promoted CD4+ T cells secretion of IL-10 and IL-21 via STAT3. Notably, TRAPs-elicited CD4+ T cells inhibited CD4+ and CD8+ effector T cell function in an IL-6- and IL-10-dependent manner and induced IL-10-producing regulatory B cells (Bregs) via IL-6, IL-10 and IL-21, thereby promoting tumor growth and metastasis. Consistently, inhibition of tumor autophagosome formation or IL-6 secretion by CD4+ T cells markedly retarded tumor growth. Furthermore, B cell or CD4+ T cell depletion impeded tumor growth by increasing effector T cell function. CONCLUSIONS HSP90α on the surface of TRAPs programs the immunosuppressive functions of CD4+ T cells to promote tumor growth and metastasis. TRAPs or their membrane-bound HSP90α represent important therapeutic targets to reverse cancer-associated immunosuppression and improve immunotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Chen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Peng-Cheng Li
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Rong Gao
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Fa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Tian-Yu Zhang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang Huang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang-Yuan Wu
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Xi-Long Ou
- Department of Gastroenterology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jin-Ping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xue-Jun Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Detroit, MI, 48201, USA.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yun-Lang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|